Skip to main content
Log in

Adipocyte and Cell Biology

Testosterone is sequestered in dysfunctional adipose tissue, modifying androgen-responsive genes

  • Article
  • Published:
International Journal of Obesity Submit manuscript

Abstract

Background/objective

The recognized association between male hypogonadism and obesity has multifactorial implications on adipose tissue (AT) physiology. The fat solubility of testosterone (T) suggests a sequestration process in fat depots, leading to reduced circulating levels of T in obesity. Several evidence suggest that steroids play a two-sided inhibitory role on adipogenesis by locally decreasing lipid accumulation and by stimulating lipolysis. The current study investigates T trafficking and activity in dysfunctional AT.

Subjects/methods

Samples of subcutaneous AT (SAT) were obtained from explants from lipoaspirate plastic surgery in six obese and six normal weight male patients. Experimental procedures on both SAT explants and insulin-resistant (IR) 3T3-L1 adipocytes were performed, including real-time PCR and mass-spectrometry quantification.

Results

A significant deregulation of gene responsiveness to androgens in IR cells and obese SAT was observed (all p < 0.05), together with reduced T release after adrenergic stimulation (−10% compared with −55% in lean SAT, p = 0.021). Higher concentrations of intracellular T and estradiol in obese SAT were also observed (2.4 vs. 1.3 ng/g, p = 0.013 and 0.075 vs. 0.22 ng/g, p = 0.004, respectively). Testosterone accumulation resulted in even lower expression in androgen-responsive genes involved in lipolytic and anti-adipogenic pathways from both in vitro and ex vivo experiments.

Conclusions

These results suggest an altered response of dysfunctional fat cells to testosterone stimulation, which normally favors lipolysis and induces an anti-adipogenic effect. The considerable reduction of lipolytic T release after adrenergic stimulation in obese SAT contributes to AT dysfunction, in a feedforward loop further reducing T levels in obese hypogonadal males.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Testosterone cromatography in adipose tissue.
Fig. 2: Testosterone content and release from ex-vivo and in vitro samples.
Fig. 3: Gene expression in human samples of subcutaneous adipose tissue from lean (empty bars) and obese (gray bars) subjects.
Fig. 4: Gene expression in normal (empty bars) and insulin-resistant (gray bars) 3T3-L1 adipocytes.

Similar content being viewed by others

References

  1. Zitzmann M. Testosterone deficiency, insulin resistance and the metabolic syndrome. Nat Rev Endocrinol. 2009;5:673–81.

    Article  CAS  Google Scholar 

  2. Mauvais-Jarvis F. Estrogen and androgen receptors: regulators of fuel homeostasis and emerging targets for diabetes and obesity. Trends Endocrinol Metab. 2011;22:24–33.

    Article  CAS  Google Scholar 

  3. Basaria S, Lieb J, Tang AM, DeWeese T, Carducci M, Eisenberger M, et al. Long-term effects of androgen deprivation therapy in prostate cancer patients. Clin Endocrinol. 2002;56:779–86.

    Article  CAS  Google Scholar 

  4. Zitzmann M, Faber S, Nieschlag E. Association of specific symptoms and metabolic risks with serum testosterone in older men. J Clin Endocrinol Metab. 2006;91:4335–43.

    Article  CAS  Google Scholar 

  5. Mårin P. Testosterone and regional fat distribution. Obes Res. 1995;3 Suppl 4:609S–12S.

    Article  Google Scholar 

  6. Wu FCW, Tajar A, Pye SR, Silman AJ, Finn JD, O’Neill TW, et al. Hypothalamic-pituitary-testicular axis disruptions in older men are differentially linked to age and modifiable risk factors: the European male aging study. J Clin Endocrinol Metab. 2008;93:2737–45.

    Article  CAS  Google Scholar 

  7. Rastrelli G, Carter EL, Ahern T, Finn JD, Antonio L, O’Neill TW, et al. Development of and recovery from secondary hypogonadism in aging men: prospective results from the EMAS. J Clin Endocrinol Metab. 2015;100:3172–82.

    Article  CAS  Google Scholar 

  8. Hammoud A, Gibson M, Hunt SC, Adams TD, Carrell DT, Kolotkin RL, et al. Effect of Roux-en-Y gastric bypass surgery on the sex steroids and quality of life in obese men. J Clin Endocrinol Metab. 2009;94:1329–32.

    Article  CAS  Google Scholar 

  9. O’Reilly MW, House PJ, Tomlinson JW. Understanding androgen action in adipose tissue. J Steroid Biochem Mol Biol. 2014;143:277–84.

  10. Blouin K, Boivin A, Tchernof A. Androgens and body fat distribution. J Steroid Biochem Mol Biol. 2008;108:272–80.

    Article  CAS  Google Scholar 

  11. Zhang Y, Calvo E, Martel C, Luu-The V, Labrie F, Tchernof A. Response of the adipose tissue transcriptome to dihydrotestosterone in mice. Physiol Genomics. 2008;35:254–61.

    Article  CAS  Google Scholar 

  12. Lafontan M, Sengenes C, Galitzky J, Berlan M, De Glisezinski I, Crampes F, et al. Recent developments on lipolysis regulation in humans and discovery of a new lipolytic pathway. Int J Obes Relat Metab Disord. 2000;24 Suppl 4:S47–52.

    Article  CAS  Google Scholar 

  13. Ryden M, Backdahl J, Petrus P, Thorell A, Gao H, Coue M, et al. Impaired atrial natriuretic peptide-mediated lipolysis in obesity. Int J Obes. 2016;40:714–20.

  14. Reynisdottir S, Wahrenberg H, Carlström K, Rössner S, Arner P. Catecholamine resistance in fat cells of women with upper-body obesity due to decreased expression of beta 2-adrenoceptors. Diabetologia. 1994;37:428–35.

    Article  CAS  Google Scholar 

  15. Di Nisio A, De Toni L, Sabovic I, Rocca MS, De Filippis V, Opocher G, et al. Impaired release of vitamin D in dysfunctional adipose tissue: new cues on vitamin D supplementation in obesity. J Clin Endocrinol Metab. 2017;102:2564–74.

    Article  Google Scholar 

  16. Langin D, Dicker A, Hoffstedt J, Mairal A, Arner E, Sicard A, et al. Obesity defects of adipose lipase. Diabetes. 2005;54:3190–97.

  17. Bélanger C, Luu-The V, Dupont P, Tchernof A. Adipose tissue intracrinology: potential importance of local androgen/estrogen metabolism in the regulation of adiposity. Horm Metab Res. 2002;34:737–45.

    Article  Google Scholar 

  18. Bélanger C, Hould F-S, Lebel S, Biron S, Brochu G, Tchernof A. Omental and subcutaneous adipose tissue steroid levels in obese men. Steroids. 2006;71:674–82.

    Article  Google Scholar 

  19. O’Reilly MW, Kempegowda P, Walsh M, Taylor AE, Manolopoulos KN, Allwood JW, et al. AKR1C3-mediated adipose androgen generation drives lipotoxicity in women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2017;102:3327–39.

    Article  Google Scholar 

  20. Drincic AT, Armas LAG, Van Diest EE, Heaney RP. Volumetric dilution, rather than sequestration best explains the low vitamin D Status of obesity. Obesity. 2012;20:1444–8.

  21. George AM, Jones PM. Perspectives on the structure–function of ABC transporters: the switch and constant contact models. Prog Biophys Mol Biol. 2012;109:95–107.

    Article  CAS  Google Scholar 

  22. Tarling EJ, de Aguiar Vallim TQ, Edwards PA. Role of ABC transporters in lipid transport and human disease. Trends Endocrinol Metab. 2013;24:342–50.

    Article  CAS  Google Scholar 

  23. Sivils JC, Gonzalez I, Bain LJ. Mice lacking Mrp1 have reduced testicular steroid hormone levels and alterations in steroid biosynthetic enzymes. Gen Comp Endocrinol. 2010;167:51–9.

  24. Dankers ACA, Roelofs MJE, Piersma AH, Sweep FCGJ, Russel FGM, van den Berg M, et al. Endocrine disruptors differentially target ATP-binding cassette transporters in the blood-testis barrier and affect Leydig cell testosterone secretion in vitro. Toxicol Sci. 2013;136:382–91.

    Article  CAS  Google Scholar 

  25. Deslypere JP, Verdonck L, Vermeulen A. Fat tissue: a steroid reservoir and site of steroid metabolism. J Clin Endocrinol Metab. 1985;61:564–70.

    Article  CAS  Google Scholar 

  26. Joyner J, Hutley L, Cameron D. Intrinsic regional differences in androgen receptors and dihydrotestosterone metabolism in human preadipocytes. Horm Metab Res. 2002;34:223–8.

    Article  CAS  Google Scholar 

  27. Blouin K, Richard C, Brochu G, Hould FS, Lebel S, Marceau S, et al. Androgen inactivation and steroid-converting enzyme expression in abdominal adipose tissue in men. J Endocrinol. 2006;191:637–49.

    Article  CAS  Google Scholar 

  28. Zhang J, Hupfeld CJ, Taylor SS, Olefsky JM, Tsien RY. Insulin disrupts beta-adrenergic signalling to protein kinase A in adipocytes. Nature. 2005;437:569–73.

    Article  CAS  Google Scholar 

  29. Arner P, Langin D. Lipolysis in lipid turnover, cancer cachexia, and obesity-induced insulin resistance. Trends Endocrinol Metab. 2014;25:255–62.

    Article  CAS  Google Scholar 

  30. Blouin K, Nadeau M, Perreault M, Veilleux A, Drolet R, Marceau P, et al. Effects of androgens on adipocyte differentiation and adipose tissue explant metabolism in men and women. Clin Endocrinol. 2010;72:176–88.

    Article  CAS  Google Scholar 

  31. Singh R, Artaza JN, Taylor WE, Gonzalez-Cadavid NF, Bhasin S. Androgens stimulate myogenic differentiation and inhibit adipogenesis in C3H 10T1/2 pluripotent cells through an androgen receptor-mediated pathway. Endocrinology. 2003;144:5081–8.

    Article  CAS  Google Scholar 

  32. Hoffstedt J, Arner P, Schalling M, Pedersen NL, Sengul S, Ahlberg S, et al. A common hormone-sensitive lipase i6 gene polymorphism is associated with decreased human adipocyte lipolytic function. Diabetes. 2001;50:2410–3.

    Article  CAS  Google Scholar 

  33. Holland AM, Roberts MD, Mumford PW, Mobley CB, Kephart WC, Conover CF, et al. Testosterone inhibits expression of lipogenic genes in visceral fat by an estrogen-dependent mechanism. J Appl Physiol. 2016;121:792–805.

    Article  Google Scholar 

  34. Anderson LA, McTernan PG, Harte AL, Barnett AH, Kumar S. The regulation of HSL and LPL expression by DHT and flutamide in human subcutaneous adipose tissue. Diabetes Obes Metab. 2002;4:209–13.

    Article  CAS  Google Scholar 

  35. Dieudonné M-N, Sammari A, Dos Santos E, Leneveu M-C, Giudicelli Y, Pecquery R. Sex steroids and leptin regulate 11β-hydroxysteroid dehydrogenase I and P450 aromatase expressions in human preadipocytes: sex specificities. J Steroid Biochem Mol Biol. 2006;99:189–96.

    Article  Google Scholar 

  36. Morris PG, Hudis CA, Giri D, Morrow M, Falcone DJ, Zhou XK, et al. Inflammation and increased aromatase expression occur in the breast tissue of obese women with breast cancer. Cancer Prev Res. 2011;4:1021–9.

    Article  CAS  Google Scholar 

  37. Subbaramaiah K, Morris PG, Zhou XK, Morrow M, Du B, Giri D, et al. Increased Levels of COX-2 and prostaglandin E 2 contribute to elevated aromatase expression in inflamed breast tissue of obese women. Cancer Discov. 2012;2:356–65.

    Article  CAS  Google Scholar 

  38. Polari L, Yatkin E, Martínez Chacón MG, Ahotupa M, Smeds A, Strauss L, et al. Weight gain and inflammation regulate aromatase expression in male adipose tissue, as evidenced by reporter gene activity. Mol Cell Endocrinol. 2015;412:123–30.

  39. Calle EE, Kaaks R. Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer. 2004;4:579–91.

    Article  CAS  Google Scholar 

  40. Finkelstein JS, Lee H, Burnett-Bowie SAM, Pallais JC, Yu EW, Borges LF, et al. Gonadal steroids and body composition, strength, and sexual function in men. N Engl J Med. 2013;369:1011–22.

  41. Colleluori G, Aguirre LE, Qualls C, Chen R, Napoli N, Villareal DT, et al. Adipocytes ESR1 expression, body fat and response to testosterone therapy in hypogonadal men vary according to estradiol levels. Nutrients. 2018;10:1226.

  42. Aversa A, Caprio M, Antelmi A, Armani A, Brama M, Greco EA, et al. Exposure to phosphodiesterase type 5 inhibitors stimulates aromatase expression in human adipocytes in vitro. J Sex Med. 2011;8:696–704.

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carlo Foresta.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Di Nisio, A., Sabovic, I., De Toni, L. et al. Testosterone is sequestered in dysfunctional adipose tissue, modifying androgen-responsive genes. Int J Obes 44, 1617–1625 (2020). https://doi.org/10.1038/s41366-020-0568-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41366-020-0568-9

  • Springer Nature Limited

This article is cited by

Navigation